bims-cepepe Biomed News
on Cell-penetrating peptides
Issue of 2025–01–12
thirteen papers selected by
Henry Lamb, Queensland University of Technology



  1. J Chem Inf Model. 2025 Jan 08.
      Language modeling applied to biological data has significantly advanced the prediction of membrane penetration for small-molecule drugs and natural peptides. However, accurately predicting membrane diffusion for peptides with pharmacologically relevant modifications remains a substantial challenge. Here, we introduce PeptideCLM, a peptide-focused chemical language model capable of encoding peptides with chemical modifications, unnatural or noncanonical amino acids, and cyclizations. We assess this model by predicting membrane diffusion of cyclic peptides, demonstrating greater predictive power than existing chemical language models. Our model is versatile and can be extended beyond membrane diffusion predictions to other target values. Its advantages include the ability to model macromolecules using chemical string notation, a largely unexplored domain, and a simple, flexible architecture that allows for adaptation to any peptide or other macromolecule data set.
    DOI:  https://doi.org/10.1021/acs.jcim.4c01441
  2. Brief Bioinform. 2024 Nov 22. pii: bbae714. [Epub ahead of print]26(1):
      The unique cyclic structure of cyclic peptides grants them remarkable stability and bioactivity, making them powerful candidates for treating various diseases. However, the lack of standardized tools for cyclic peptide data has hindered their potential in today's artificial intelligence-driven efficient drug design landscape. To bridge this gap, here we introduce a Python package named cyclicpeptide specifically for cyclic peptide drug design. This package provides standardized tools such as Structure2Sequence, Sequence2Structure, and format transformation to process, convert, and standardize cyclic peptide structure and sequence data. Additionally, it includes GraphAlignment for cyclic peptide-specific alignment and search and PropertyAnalysis to enhance the understanding of their drug-like properties and potential applications. This comprehensive suite of tools aims to streamline the integration of cyclic peptides into modern drug discovery pipelines, accelerating the development of cyclic peptide-based therapeutics.
    Keywords:  artificial intelligence; bioinformatics tool; cyclic peptide; drug design
    DOI:  https://doi.org/10.1093/bib/bbae714
  3. Bioorg Med Chem. 2024 Nov 27. pii: S0968-0896(24)00431-0. [Epub ahead of print]119 118017
      Interleukin-11 (IL-11), a member of the IL-6 cytokine family, has potential pro-inflammatory and pro-fibrotic roles in pulmonary, hepatic, cardiovascular, renal and intestinal disease pathogenesis, including oncogenesis. The potential for therapeutic intervention in these disease spaces has therefore made the IL-11 signaling axis an attractive target in drug discovery, and antibody inhibitors of IL-11 signaling are currently under evaluation in Phase I/II clinical trials. While lower molecular weight small molecule and peptide inhibitors may offer the potential for improved tissue penetration, developability and manufacturing cost compared with a protein therapeutic, reports of such chemical matter in the literature are limited. In this work, a series of cyclic peptides derived from phage display biopanning campaigns against both IL-11 and its cognate receptor IL-11Rα are presented. The most active IL-11 binder (peptide 4, KD 140 nM) exhibited inhibition of IL-11/IL-11Rα dimerization in a biochemical AlphaLISA assay (Ki 300 nM), and alanine scanning was carried out on this sequence to identify residues important for target binding and inhibitory activity. Further structural optimization yielded lead peptide 15 (Ki 180 nM), which exhibited at least 70-fold greater activity than IL-11 inhibitors previously reported in the literature. The de novo peptide macrocycles presented serve as a robust starting point for development of therapeutic inhibitors of the IL-11/IL-11Rα interaction.
    Keywords:  AlphaLISA; Antagonist; Cytokine; IL-11; Inhibitor; Peptide; Phage display
    DOI:  https://doi.org/10.1016/j.bmc.2024.118017
  4. ArXiv. 2025 Jan 01. pii: arXiv:2412.17780v3. [Epub ahead of print]
      Peptide therapeutics, a major class of medicines, have achieved remarkable success across diseases such as diabetes and cancer, with landmark examples such as GLP-1 receptor agonists revolutionizing the treatment of type-2 diabetes and obesity. Despite their success, designing peptides that satisfy multiple conflicting objectives, such as target binding affinity, solubility, and membrane permeability, remains a major challenge. Classical drug development and structure-based design are ineffective for such tasks, as they fail to optimize global functional properties critical for therapeutic efficacy. Existing generative frameworks are largely limited to continuous spaces, unconditioned outputs, or single-objective guidance, making them unsuitable for discrete sequence optimization across multiple properties. To address this, we present PepTune, a multi-objective discrete diffusion model for the simultaneous generation and optimization of therapeutic peptide SMILES. Built on the Masked Discrete Language Model (MDLM) framework, PepTune ensures valid peptide structures with state-dependent masking schedules and penalty-based objectives. To guide the diffusion process, we propose a Monte Carlo Tree Search (MCTS)-based strategy that balances exploration and exploitation to iteratively refine Pareto-optimal sequences. MCTS integrates classifier-based rewards with search-tree expansion, overcoming gradient estimation challenges and data sparsity inherent to discrete spaces. Using PepTune, we generate diverse, chemically-modified peptides optimized for multiple therapeutic properties, including target binding affinity, membrane permeability, solubility, hemolysis, and non-fouling characteristics on various disease-relevant targets. In total, our results demonstrate that MCTS-guided discrete diffusion is a powerful and modular approach for multi-objective sequence design in discrete state spaces.
  5. Angew Chem Int Ed Engl. 2025 Jan 07. e202424076
      Domain antibodies such as monobodies provide an attractive immunoglobin fold for evolving high-affinity protein binders targeting the intracellular proteins implicated in cell signalling. However, it remains a challenge to endow cell permeability to these small and versatile protein binders. Here, we report a streamlined approach combining orthogonal crosslinking afforded by a genetically encoded β-lactam-lysine (BeLaK) and genetic supercharging to generate cell-penetrating monobodies. When introduced to the N-terminal β-strand of a series of supercharged monobodies, BeLaK enabled efficient inter-strand crosslinking with the neighbouring lysine. Compared to its non-crosslinked counterpart, a BeLaK-crosslinked, +18-charged monobody exhibited enhanced thermostability and greater cellular uptake at 40 nM. Moreover, this structurally rigidified, supercharged monobody inhibited ERK1/2 phosphorylation in KYSE-520 esophageal cancer cell line at sub-micromolar concentration, indicating significant endosomal escape after endocytosis. Together, the discovery of this BeLaK-encoded, rigidified immunoglobin fold should facilitate the design of cell-penetrating monobodies targeting intracellular signalling proteins.
    Keywords:  Domain antibody; Orthogonal crosslinking; Proximity-driven reaction; beta-Lactam; genetic code expansion
    DOI:  https://doi.org/10.1002/anie.202424076
  6. ACS Nano. 2025 Jan 06.
      The tumor-specific efficacy of the most current anticancer therapeutic agents, including antibody-drug conjugates (ADCs), oligonucleotides, and photosensitizers, is constrained by limitations such as poor cell penetration and low drug delivery. In this study, we addressed these challenges by developing, a positively charged, amphiphilic Chlorin e6 (Ce6)-conjugated, cell-penetrating anti-PD-L1 peptide nanomedicine (CPPD1) with enhanced cell and tissue permeability. The CPPD1 molecule, a bioconjugate of a hydrophobic photosensitizer and strongly positively charged programmed cell death-ligand 1 (PD-L1) binding cell-penetrating peptide (CPP), is capable of self-assembling into nanoparticles with an average size of 199 nm in aqueous solution without the need for any carriers. These carrier-free nanoparticles possess the ability to penetrate the cell membrane of cancer cells and target tumors expressing PD-L1 on their surface. Notably, CPPD1 nanoparticles effectively blocked programmed cell death-1 (PD-1)/PD-L1 interactions and reduced PD-L1 expression via lysosomal degradation. They also demonstrated the responsiveness of CPPD1 nanoparticles in photodynamic therapy (PDT) to a 635 nm laser, leading to the generation of ROS, and induction of various immunogenic cell deaths (ICD). Highly penetrating CPPD1 nanoparticles could immunogenically modulate the microenvironment of CT26 cancer and were also effective in treating abscopal metastatic tumors, addressing major limitations of traditional PDT.
    Keywords:  abscopal effect; anti-PD-L1; cell-penetrating peptide; immunogenic cell death; photodynamic therapy; self-assembly
    DOI:  https://doi.org/10.1021/acsnano.4c16128
  7. bioRxiv. 2024 Dec 20. pii: 2024.12.16.628822. [Epub ahead of print]
      T cell receptor (TCR) mimics offer a promising platform for tumor-specific targeting of peptide-MHC in cancer immunotherapy. Here, we designed a de novo α-helical TCR mimic (TCRm) specific for the NY-ESO-1 peptide presented by HLA-A * 02, achieving high on-target specificity with nanomolar affinity (K d = 9.5 nM). The structure of the TCRm/pMHC complex at Å resolution revealed a rigid TCR-like docking mode with an unusual degree of focus on the up-facing NY-ESO-1 side chains, suggesting the potential for reduced off-target reactivity. Indeed, a structure-informed in silico screen of 14,363 HLA-A * 02 peptides correctly predicted two off-target peptides, yet our TCRm maintained a wide therapeutic window as a T cell engager. These results represent a path for precision targeting of tumor antigens with peptide-focused α-helical TCR mimics.
    DOI:  https://doi.org/10.1101/2024.12.16.628822
  8. Bioorg Med Chem. 2024 Dec 28. pii: S0968-0896(24)00469-3. [Epub ahead of print]119 118055
      Polo-like kinase 1 (Plk1) is an important cell cycle regulator that is a recognized target for development of anti-cancer therapeutics. Plk1 is composed of a catalytic kinase domain (KD), a flexible interdomain linker and a polo-box domain (PBD). Intramolecular protein-protein interactions (PPIs) between the PBD and KD result in "auto-inhibition" that is an essential component of proper Plk1 function. Recently, we developed high-affinity PBD-binding inhibitors using a bivalent approach. These ligands contain the low-nanomolar affinity Plk1 KD-binding inhibitors BI2536 or Wortmannin tethered to the PBD-binding peptide, PLH*SpT (H* represents a -(CH2)8Ph group on the histidine side chain π-nitrogen). Due to the extremely high affinity of these bivalent inhibitors, to avoid bottoming out in competitive binding assays, it was necessary to use PLH*SpT in the affinity probe. As reported herein, we have developed fluorescence polarization assays using a new fluorescent probe based on the Plk1 PBD-binding peptide, FDPPLHSpTA. We applied the assay to evaluate the affinities of bivalent inhibitors that possess a variety of PBD-binding peptides having much lower PBD-affinities than PLH*SpT. Tethering BI2536 in these bivalent inhibitors resulted in significant affinity enhancements as compared to the parent monovalent peptides.
    Keywords:  Bivalent inhibitor; Fluorescence Polarization (FP) assay; Phosphorylated threonine (pT); Polo-box domain (PBD); Polo-like kinase 1 (Plk1)
    DOI:  https://doi.org/10.1016/j.bmc.2024.118055
  9. Org Biomol Chem. 2025 Jan 07.
      Macrocyclic peptides containing a thiazole heterocycle exhibit fascinating properties in natural products and future therapeutics. We report a biocompatible macrocyclisation approach facilitated by an N-terminal cysteine and C-terminal nitriles. The use of various chiral α-amino nitriles enables the incorporation of diverse hydrophobic side chains adjacent to the thiazole motif.
    DOI:  https://doi.org/10.1039/d4ob01989j
  10. Microfluid Nanofluidics. 2024 Jul;pii: 44. [Epub ahead of print]28(7):
      The blood-brain barrier (BBB) protects the brain by actively allowing the entry of ions and nutrients while limiting the passage of from toxins and pathogens. A healthy BBB has low permeability and high selectivity to maintain normal brain functions. Increased BBB permeability can result from neurological diseases and traumatic injuries. Modern engineering technologies such as microfluidics and fabrication techniques have advanced the development of BBB models to simulate the basic functions of BBB. However, the intrinsic BBB properties are difficult to replicate. Existing in vitro BBB models demonstrate inconsistent BBB permeability and selectivity due to variations in microfluidic design, cell types and arrangement, expression of tight junction (TJ) proteins, and use of shear stress. Specifically, microfluidic designs have flow channels of different sizes, complexity, topology, and modular structure. Different cell types are selected to mimic various physiological conditions. These factors make it challenging to compare results obtained using different experimental setups. This paper highlights key factors that play important roles in influencing microfluidic models and discusses how these factors contribute to permeability and selectivity of the BBB models.
    Keywords:  Blood-brain barrier; Cells; Design; Microfluidics; Permeability; Selectivity
    DOI:  https://doi.org/10.1007/s10404-024-02741-z
  11. Biomedicines. 2024 Dec 18. pii: 2873. [Epub ahead of print]12(12):
      Background: Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a growing class of natural products biosynthesized from a genetically encoded precursor peptide. RiPPs have attracted attention for the ability to generate and screen libraries of these compounds for useful biological activities. To facilitate this screening, it is useful to be able to do so with the leader peptide still present. We assessed the suitability of the microviridin family for these screening experiments by determining their activity with the leader peptide still present. Methods: Modified precursor peptides with the leader present were heterologously expressed in Escherichia coli. Their ability to inhibit elastase was tested with a fluorogenic substrate. HPLC was used to monitor degradation of the modified precursor peptides by elastase. SDS-PAGE was used to determine the ability of immobilized modified precursor peptide to pull down elastase. Results: We found that the fully modified precursor peptide of microviridin B can inhibit the serine protease elastase with a low nanomolar IC50, and that the fully modified precursor with an N-terminal His-tag can mediate interactions between elastase and Ni-NTA resin, all indicating leader peptide removal is not necessary for microviridins to bind their target proteases. Additionally, we found that a bicyclic variant was able to inhibit elastase with the leader peptide still present, although with a roughly 100-fold higher IC50 and being subject to hydrolysis by elastase. Conclusions: These results open a pathway to screening libraries of microviridin variants for improved protease inhibition or other characteristics that can serve as, or as inspirations for, new pharmaceuticals.
    Keywords:  RiPP; elastase; microviridin
    DOI:  https://doi.org/10.3390/biomedicines12122873
  12. Pharmaceutics. 2024 Dec 19. pii: 1613. [Epub ahead of print]16(12):
      Background/Objectives: Here, we report on the synthesis and biological evaluation of a novel peptide-drug conjugate, P6-SN38, which consists of the EGFR-specific short cyclic peptide, P6, and the Topo I inhibitor SN38, which is a bioactive metabolite of the anticancer drug irinotecan. Methods: SN38 is attached to the peptide at position 20 of the E ring's tertiary hydroxyl group via a mono-succinate linker. Results: The developed peptide-drug conjugate (PDC) exhibited sub-micromolar anticancer activity on EGFR-positive (EGFR+) cell lines but no effect on EGFR-negative (EGFR-) cells. In vivo studies have shown that this PDC specifically accumulates in EGFR+ non-small cell lung cancer (NSCLC) xenografts and presents superior anticancer activity compared to the EGFR-specific antibody cetuximab (ErbituxTM) and free SN38. The 10 mg/kg dose of P6-SN38 in a side-by-side EGFR+/EGFR- xenograft shows eradication of the EGFR+ tumor with good tolerance, but no inhibition of tumor growth of the EGFR- counterpart. Conclusions: The PDC examined in this study was proven to be highly efficient for NSCLC, broadening its utilization for targeted cancer therapy in EGFR overexpressed cancers.
    Keywords:  EGFR; NSCLC; SN38; peptide–drug conjugate; targeted cancer therapy; topo I inhibitor
    DOI:  https://doi.org/10.3390/pharmaceutics16121613
  13. J Med Chem. 2025 Jan 04.
      Radiolabeled peptides are vital for positron emission tomography (PET) imaging, yet the 18F-labeling peptides remain challenging due to harsh conditions and time-consuming premodification requirements. Herein, we developed a novel vinyltetrazine-mediated bioorthogonal approach for highly efficient 18F-radiolabeling of a native peptide under mild conditions. This approach enabled radiosynthesis of various tumor-targeting PET tracers, including targeting the neurofibromin receptor (18F-P10a), the integrin αvβ3 (18F-P12a), and the platelet-derived growth factor receptor β (18F-ZPDGFRβ), with a radiochemical yield exceeding 90%. Preliminary evaluations revealed excellent hydrophilicity across these tracers, with 18F-P12a effectively visualizing integrin αvβ3 expression (tumor uptake: 1.57 ± 0.54%ID/g at 2 h). Additionally, we explored the potential for development of PET/near-infrared (NIR) dual-labeling agents using this method. The dual-modality agent 18F-Cy5-P12d enables specificity and colocalized imaging integrin αvβ3 expression (tumor uptake: 1.35 ± 0.24%ID/g at 2 h). Overall, this strategy offers a versatile platform for peptide radiolabeling and dual-modality agent development.
    DOI:  https://doi.org/10.1021/acs.jmedchem.4c02165